1
|
Dessenne C, Mariller C, Vidal O, Huvent I, Guerardel Y, Elass-Rochard E, Rossez Y. Glycan-mediated adhesion mechanisms in antibiotic-resistant bacteria. BBA ADVANCES 2025; 7:100156. [PMID: 40207210 PMCID: PMC11979486 DOI: 10.1016/j.bbadva.2025.100156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 04/11/2025] Open
Abstract
Bacterial adhesins play a central role in host-pathogen interactions, with many specifically targeting glycans to mediate bacterial colonization, influence infection dynamics, and evade host immune responses. In this review, we focus on bacterial pathogens identified by the World Health Organization as critical threats to public health and in urgent need of new treatments. We summarize glycoconjugate targets identified in the literature across 19 bacterial genera and species. This comprehensive review provides a foundation for the development of innovative therapeutic strategies to effectively combat these pathogens.
Collapse
Affiliation(s)
- Clara Dessenne
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Christophe Mariller
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Olivier Vidal
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Isabelle Huvent
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Yann Guerardel
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan
| | - Elisabeth Elass-Rochard
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| | - Yannick Rossez
- Université Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Lille, France
| |
Collapse
|
2
|
Trbovc A, Pušnik M, Šteferl T, Hajdinjak M, Štukelj M. Cross-Sectional Study: Assessing the Presence of Stx2e-Producing E. coli Virotypes in Samples of Oral Fluid of Growers and Fatteners. Pathogens 2025; 14:261. [PMID: 40137746 PMCID: PMC11944655 DOI: 10.3390/pathogens14030261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Edema disease is a multifactorial infectious disease caused by specific E. coli virotypes possessing fimbriae F18 and toxin Stx2e that cause significant losses in the post-weaning period. The aim of this study was to assess the presence of Stx2e-producing E. coli verotypes in Slovenian commercial pig farms in relation to the biosecurity and technological measures undertaken by the owners. Samples of oral fluid were collected from growers and fatteners at 5-6 weeks, 7-8 weeks, 12 weeks and 14 weeks of age on 37 commercial pig farms, using the Verocheck® diagnostic kit for the real-time PCR detection of Stx2e. The results of RT-PCR and the questionnaire were statistically analyzed. The prevalence of E. coli strains producing Stx2e was 64.9%. Statistically significant association between the prevalence of Stx2e producing E. coli strains and the type of the farm and feed origin was proved. No association was found between prevalence and farm size, presence of quarantine or previous outbreaks of edema disease. None of the studied age groups showed a statistically significant dominance in prevalence compared to other age groups, which contradicts the current theoretical data. Further studies are needed to estimate the proportion of Stx2e produced by the EDEC pathotype compared to other E. coli strains.
Collapse
Affiliation(s)
- Ana Trbovc
- Clinic for Reproduction and Large Animals, Clinic for Ruminants and Pigs, Veterinary Faculty, University of Ljubljana, Cesta v Mestni log 47, 1000 Ljubljana, Slovenia; (A.T.)
| | - Matevž Pušnik
- Clinic for Reproduction and Large Animals, Clinic for Ruminants and Pigs, Veterinary Faculty, University of Ljubljana, Cesta v Mestni log 47, 1000 Ljubljana, Slovenia; (A.T.)
| | - Tim Šteferl
- Clinic for Reproduction and Large Animals, Clinic for Ruminants and Pigs, Veterinary Faculty, University of Ljubljana, Cesta v Mestni log 47, 1000 Ljubljana, Slovenia; (A.T.)
| | - Melita Hajdinjak
- Laboratory of Applied Mathematics and Statistics, Faculty of Electrical Engineering, University of Ljubljana, Tržaška cesta 25, 1000 Ljubljana, Slovenia;
| | - Marina Štukelj
- Clinic for Reproduction and Large Animals, Clinic for Ruminants and Pigs, Veterinary Faculty, University of Ljubljana, Cesta v Mestni log 47, 1000 Ljubljana, Slovenia; (A.T.)
| |
Collapse
|
3
|
Wu Z, Li M, Wu J, Jin S, Xu Y, Jin J, Wu Y. Characterization of the molecular role that ST3GAL1 plays in porcine susceptibility to E. coli F18 infection. Int J Biol Macromol 2024; 276:133959. [PMID: 39029847 DOI: 10.1016/j.ijbiomac.2024.133959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/09/2024] [Accepted: 07/16/2024] [Indexed: 07/21/2024]
Abstract
Escherichia coli F18 (E. coli F18) is the main cause of bacterial diarrhea in piglets. Previous transcriptome reported that ST3GAL1 was associated to E. coli F18 infection. However, its role in mediating the resistance to E. coli F18 remains elusive. Here, we revealed that the downregulation of ST3GAL1 expression contributed to the enhancement of E. coli F18 resistance in IPEC-J2 cells. Bisulfite sequencing identified 26 methylated CpG sites in the ST3GAL1 core promoter. Among these, the ST3GAL1 mRNA levels significantly correlated with methylation levels of the mC-8 site in the specificity protein 1 (SP1) transcription factor (P < 0.01). Interestingly, ST3GAL1 expression may enhances the immune response by activating TLRs signaling, meanwhile decreases the production of the E. coli F18 receptor by inhibiting glycosphingolipid biosynthesis signaling, thereby leading to enhance the resistance to E. coli F18 infection. Besides, low ST3GAL1 expression may increase E. coli resistance by reducing sialylation. Together, these results support the status of ST3GAL1 as a viable target for efforts to modulate E. coli F18 susceptibility, offering a theoretical foundation for the use of this gene as a key biomarker for molecular breeding to improve porcine disease resistance.
Collapse
Affiliation(s)
- Zhengchang Wu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China; College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Meiqi Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Jiayun Wu
- Jiangsu Agri-animal Husbandry Vocational College, Taizhou 225300, China
| | - Shuting Jin
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yifan Xu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Jian Jin
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China.
| | - Yanqing Wu
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, Jiangsu, China.
| |
Collapse
|
4
|
Kim K, Jinno C, Li X, Bravo D, Cox E, Ji P, Liu Y. Impact of an oligosaccharide-based polymer on the metabolic profiles and microbial ecology of weanling pigs experimentally infected with a pathogenic E. coli. J Anim Sci Biotechnol 2024; 15:1. [PMID: 38169416 PMCID: PMC10759389 DOI: 10.1186/s40104-023-00956-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 10/29/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Our previous study has reported that supplementation of oligosaccharide-based polymer enhances gut health and disease resistance of pigs infected with enterotoxigenic E. coli (ETEC) F18 in a manner similar to carbadox. The objective of this study was to investigate the impacts of oligosaccharide-based polymer or antibiotic on the host metabolic profiles and colon microbiota of weaned pigs experimentally infected with ETEC F18. RESULTS Multivariate analysis highlighted the differences in the metabolic profiles of serum and colon digesta which were predominantly found between pigs supplemented with oligosaccharide-based polymer and antibiotic. The relative abundance of metabolic markers of immune responses and nutrient metabolisms, such as amino acids and carbohydrates, were significantly differentiated between the oligosaccharide-based polymer and antibiotic groups (q < 0.2 and fold change > 2.0). In addition, pigs in antibiotic had a reduced (P < 0.05) relative abundance of Lachnospiraceae and Lactobacillaceae, whereas had greater (P < 0.05) Clostridiaceae and Streptococcaceae in the colon digesta on d 11 post-inoculation (PI) compared with d 5 PI. CONCLUSIONS The impact of oligosaccharide-based polymer on the metabolic and microbial profiles of pigs is not fully understood, and further exploration is needed. However, current research suggest that various mechanisms are involved in the enhanced disease resistance and performance in ETEC-challenged pigs by supplementing this polymer.
Collapse
Affiliation(s)
- Kwangwook Kim
- Department of Animal Science, University of California, Davis, CA, 95616, USA
- Present Affiliation: Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA
| | - Cynthia Jinno
- Department of Animal Science, University of California, Davis, CA, 95616, USA
- Present Affiliation: Cedars-Sinai Medical Center, Los Angeles, CA, 90084, USA
| | - Xunde Li
- School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - David Bravo
- Pancosma|ADM, 1180, Rolle, Switzerland
- Present Affiliation: Nutreco Exploration, Nutreco, The Netherlands
| | - Eric Cox
- Department of Virology, Parasitology and Immunology, Ghent University, 9000, Ghent, Belgium
| | - Peng Ji
- Department of Nutrition, University of California, Davis, CA, 95616, USA
| | - Yanhong Liu
- Department of Animal Science, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
5
|
Berger PI, Hermanns S, Kerner K, Schmelz F, Schüler V, Ewers C, Bauerfeind R, Doherr MG. Cross-sectional study: prevalence of oedema disease Escherichia coli (EDEC) in weaned piglets in Germany at pen and farm levels. Porcine Health Manag 2023; 9:49. [PMID: 37885038 PMCID: PMC10601234 DOI: 10.1186/s40813-023-00343-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND Escherichia coli bacteria capable of producing the toxin Stx2e and possessing F18-fimbriae (edema disease E. coli, EDEC) are considered causative agents of porcine oedema disease. This disease, which usually occurs in piglets shortly after weaning, has a high lethality in affected animals and can lead to high economic losses in piglet rearing. The aim of this cross-sectional field study was to determine the prevalence of EDEC in weaned piglets in Germany at pen and farm levels. RESULTS Ninety-nine farms with unknown history of infections with shigatoxin-producing E. coli (STEC) and oedema disease were sampled. On each farm, up to five pens were selected for sampling (n = 481). The piglets in these pens were at an age 1-3 weeks after weaning. Single faecal samples (n = 2405) and boot swabs (n = 479) were collected from the floor. On 50 farms, cotton ropes were additionally used to collect oral fluid samples (n = 185) and rope wash out samples (n = 231) from the selected pens. All samples were analyzed by bacterial culture combined with a duplex PCR for the presence of the corresponding genes stx2e and fedA (major subunit protein of F18 fimbriae). In addition, whole DNA specimens extracted from boot swabs, oral fluid samples, and rope wash out samples were directly examined by duplex PCR for DNA of stx2e and fedA. A pen was classified as positive if at least one of the samples, regardless of the technique, yielded a positive result in the PCR, and farms were considered positive if at least one pen was classified as positive. Overall, genes stx2e and fedA were found simultaneously in 24.9% (95% CI 22.1-29.1%) of sampled pens and in 37.4% (95% CI 27.9-47.7%) of sampled farms. Regardless of the presence of F18-fimbriae, Escherichia coli encoding for Stx2e (STEC-2e) were found in 35.1% (95% CI 31.0-39.1%) of the pens and 53.5% (95% CI 44.4-63.6%) of the farms sampled. CONCLUSIONS Escherichia coli strains considered capable to cause oedema disease in swine (EDEC) are highly prevalent in the surveyed pig producing farms in Germany. Due to intermittent shedding of EDEC and a potentially low within-farm prevalence, we recommend a combination of different sampling techniques for EDEC monitoring at pen and farm levels. Further studies are needed to understand which STEC-2e strains really pose the risk of causing severe porcine disease.
Collapse
Affiliation(s)
- Pia I Berger
- Institute of Veterinary Epidemiology and Biostatistics, Freie Universität Berlin, Berlin, Germany.
| | - Steffen Hermanns
- Institute for Hygiene and Infectious Diseases of Animals, Justus Liebig University, Giessen, Germany
| | - Katharina Kerner
- Institute for Hygiene and Infectious Diseases of Animals, Justus Liebig University, Giessen, Germany
| | | | | | - Christa Ewers
- Institute for Hygiene and Infectious Diseases of Animals, Justus Liebig University, Giessen, Germany
| | - Rolf Bauerfeind
- Institute for Hygiene and Infectious Diseases of Animals, Justus Liebig University, Giessen, Germany
| | - Marcus G Doherr
- Institute of Veterinary Epidemiology and Biostatistics, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
6
|
Song D, Lee J, Kwak W, Oh H, Chang S, An J, Cho H, Park S, Jeon K, Cho J. Effects of stimbiotic supplementation on gut health, immune response, and intestinal microbiota in weaned piglets challenged with E. coli. Front Vet Sci 2023; 10:1187002. [PMID: 37538167 PMCID: PMC10394646 DOI: 10.3389/fvets.2023.1187002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/27/2023] [Indexed: 08/05/2023] Open
Abstract
In order to make piglet diets more effective, it is necessary to investigate effective methods for breaking down xylan in cereal. The objective of this study was to determine the effects of dietary stimbiotic (STB) supplementation on growth performance, intestinal morphology, immune response and intestinal microbiota in weaned piglets. A total of 24 (Duroc × Yorkshire × Landrace) weaned pigs (initial body weight of 8.01 ± 0.38 kg and 28 ± 3 d old), were assigned to 4 treatments with 6 replicates per treatment. Pigs were housed in individual pens for 17 days, including 5 days adaption period and 12 days after the first Escherichia coli (E. coli) challenge. The experiment was conducted in a 2 × 2 factorial arrangement of treatments consisting of two levels of challenge (challenge and non-challenge) and two levels of STB (0 and 0.5 g/kg diet). Supplementations of STB 0.5 g/kg improved the gain to feed ratio (G:F) (P < 0.05) in piglets challenged with shiga toxigenic E. coli (STEC). STB supplementation decreased (P < 0.05) white blood cells, neutrophils, lymphocytes, and expression levels of tumor necrosis factor-alpha and interleukin-6. Supplementation of STB improved (P < 0.05) the lymphocytes and neutrophils in piglets challenged with STEC on 12 dpi. Supplementation of STB also improved (P < 0.05) the villus height to-crypt depth ratio of ileum in piglets challenged with STEC. Supplementation of STB increased (P < 0.05) the expression levels of claudin-1 of ileum. In genus level, supplementation of STB increased (P < 0.001) the abundance of Prevotella compared to non-supplementation of STB groups in pre-inoculation period. Also, supplementation of STB decreased (P < 0.05) the abundance of Faecalibacterium and Eubacterium_coprostanoligenes_group compared to non-supplementation of STB groups in post-inoculation period. In phylum level, supplementation of STB increased (P < 0.05) the abundance of Desulfobacterota and Fibrobacterota in pre-inoculation period. E. coli challenge increased the abundance of Fibrobacterota compared to non-challenged group in post-inoculation period. In conclusion, these findings indicated that STB supplementation could alleviate a decrease of the performance, immune response, and inflammatory response in piglets induced by the STEC challenge.
Collapse
Affiliation(s)
- Dongcheol Song
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Jihwan Lee
- Department of Poultry Science, University of Georgia (UGA), Athens, GA, United States
| | - Woogi Kwak
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Hanjin Oh
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Seyeon Chang
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Jaewoo An
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Hyunah Cho
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Sehyun Park
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Kyeongho Jeon
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| | - Jinho Cho
- Department of Animal Science, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
7
|
Quintana-Hayashi MP, Zalem D, Lindén S, Teneberg S. Porcine intestinal glycosphingolipids recognized by Brachyspira hyodysenteriae. Microb Pathog 2023; 175:105961. [PMID: 36581306 DOI: 10.1016/j.micpath.2022.105961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Swine dysentery caused by Brachyspira hyodysenteriae is a disease present worldwide with an important economic impact on the farming business, resulting in an increased use of antibiotics. In the present study, we investigated the binding of B. hyodysenteriae to glycosphingolipids from porcine small intestinal epithelium in order to determine the glycosphingolipids involved in B. hyodysenteriae adhesion. Specific interactions between B. hyodysenteriae and two non-acid glycosphingolipids were obtained. These binding-active glycosphingolipids, were characterized by mass spectrometry as lactotetraosylceramide (Galβ3GlcNAcβ3Galβ4Glcβ1Cer) and the B5 glycosphingolipid (Galα3Galβ4GlcNAcβ3Galβ4Glcβ1Cer). Comparative binding studies using structurally related reference glycosphingolipids showed that B. hyodysenteriae binding to lactotetraosylceramide required an unsubstituted terminal Galβ3GlcNAc sequence, while for binding to the B5 pentaosylceramide the terminal Galα3Galβ4GlcNAc sequence is the minimum element recognized by the bacteria. Binding of Griffonia simplicifolia IB4 lectin to pig colon tissue sections from healthy control pig and B. hyodysenteriae infected pigs showed that in the healthy pigs the Galα3Gal epitope was mainly present in the lamina propria. In contrast, in four out of five pigs with swine dysentery there was an increased expression of Galα3Gal in the goblet cells and in the colonic crypts, where B. hyodysenteriae also was present. The one pig that had recovered by the time of necropsy had the Galα3Gal epitope only in the lamina propria. These data are consistent with a model where a transient increase in the carbohydrate sequence recognized by the bacteria occur in colonic mucins during B. hyodysenteriae infection, suggesting that the mucins may act as decoys contributing to clearance of the infection. These findings may lead to novel strategies for treatment of B. hyodysenteriae induced swine dysentery.
Collapse
Affiliation(s)
- Macarena P Quintana-Hayashi
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Dani Zalem
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Sara Lindén
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Susann Teneberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden.
| |
Collapse
|
8
|
Raev SA, Omwando AM, Guo Y, Raque MS, Amimo JO, Saif LJ, Vlasova AN. Glycan-mediated interactions between bacteria, rotavirus and the host cells provide an additional mechanism of antiviral defence. Benef Microbes 2022; 13:383-396. [PMID: 36239669 DOI: 10.3920/bm2022.0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Limited efficacy of rotavirus (RV) vaccines in children in developing countries and in animals remains a significant problem necessitating further search for additional approaches to control RV-associated gastroenteritis. During cell attachment and entry events, RV interacts with cell surface O-glycans including histo-blood group antigens (HBGAs). Besides modulation of the protective immunity against RV, several commensal and probiotic bacteria were shown to express HBGA-like substances suggesting that they may affect RV attachment and entry into the host cells. Moreover, some beneficial bacteria have been shown to possess the ability to bind host HBGAs via sugar specific proteins called lectins. However, limited research has been done to evaluate the effects of HBGA-expressing and/or HBGA-binding bacteria on RV infection. The aim of this study was to investigate the ability of selected commensal and probiotic bacteria to bind different RV strains via HBGAs and to block RV infection of IPEC-J2 cells. Our data indicated that Gram-negative probiotic Escherichia coli Nissle 1917 (E. coli Nissle 1917) and commensal Gram-positive (Streptococcus bovis and Bifidobacterium adolescentis) and Gram-negative (Bacteroides thetaiotaomicron, Clostridium clostridioforme and Escherichia coli G58 (E. coli G58) bacteria of swine origin expressed HBGAs which correlated with their ability to bind group A and C RVs. Additionally, Gram-positive E. coli 1917 and E. coli G58 demonstrated the ability to block RV attachment onto IPEC-J2 cells. Taken together, our results support the hypothesis that physical interactions between RVs and HBGA-expressing beneficial bacteria may limit RV replication.
Collapse
Affiliation(s)
- S A Raev
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - A M Omwando
- Department of Public Health, Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Nairobi, P.O. Box 29053, 00625 Nairobi, Kenya
| | - Y Guo
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - M S Raque
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - J O Amimo
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
- Department of Animal Production, Faculty of Veterinary Medicine, University of Nairobi, P.O. Box 29053, 00625 Nairobi, Kenya
| | - L J Saif
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| | - A N Vlasova
- Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA
| |
Collapse
|
9
|
Comprehensive Analysis Revealed the Potential Roles of N6-Methyladenosine (m6A) Mediating E. coli F18 Susceptibility in IPEC-J2 Cells. Int J Mol Sci 2022; 23:ijms232113602. [DOI: 10.3390/ijms232113602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/20/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022] Open
Abstract
Post-weaning diarrhea caused by enterotoxigenic Escherichia coli F18 (E. coli F18) causes significant economic losses for pig producers. N6-methyladenosine (m6A) is a highly abundant epitranscriptomic marker that has been found to be involved in regulating the resistance of host cells to pathogenic infection, but its potential role in E. coli F18-exposed intestinal porcine epithelial cells (IPEC-J2) remains undetermined. Here, we demonstrated that m6A and its regulators modulate E. coli F18 susceptibility. Briefly, we revealed that the Wilms’ tumor 1-associating protein (WTAP) expressions were markedly elevated in IPEC-J2 cells upon E. coli F18 exposure. WTAP are required for the regulation of E. coli F18 adhesion in IPEC-J2 cells. Additionally, WTAP knockdown significantly suppressed m6A level at N-acetyllactosaminide beta-1,6-N-acetylglucosaminyl-transferase (GCNT2) 3′UTR, resulting in the enhancement of TH N6-methyladenosine RNA binding protein 2 (YTHDF2)-mediated GCNT2 mRNA stability. Subsequently, the altered GCNT2 expressions could inhibit the glycosphingolipid biosynthesis, thus improving resistance to E. coli F18 infection in IPEC-J2. Collectively, our analyses highlighted the mechanism behind the m6A-mediated management of E. coli F18 susceptibility, which will aid in the development of novel approaches that protect against bacterial diarrhea in piglets.
Collapse
|
10
|
Lactoferrin Decreases Enterotoxigenic Escherichia coli-Induced Fluid Secretion and Bacterial Adhesion in the Porcine Small Intestine. Pharmaceutics 2022; 14:pharmaceutics14091778. [PMID: 36145526 PMCID: PMC9504966 DOI: 10.3390/pharmaceutics14091778] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/25/2022] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) infections are one of the most prevalent causes of post-weaning diarrhea in piglets, resulting in morbidity, mortality and elevated use of antibiotics. The emergence and further spread of antimicrobial resistance together with the growing demand for high quality animal protein requires the identification of novel alternatives for antimicrobials. A promising alternative is lactoferrin, as we previously showed that it can both inhibit the growth and degrade bacterial virulence factors of porcine ETEC strains in vitro. Aiming to confirm these findings in vivo, we performed a small intestinal segment perfusion experiment in piglets. Here, we showed that lactoferrin could not only decrease ETEC-induced fluid secretion, but also their ability to colonize the small intestinal epithelium. Furthermore, while ETEC infection induced pro-inflammatory cytokine mRNA expression in this experiment, lactoferrin was not able to counteract these responses. In addition, a bacterial motility assay showed that lactoferrin can reduce the motility of ETEC. Our findings further support the use of lactoferrin as an alternative for antimicrobials and also show its potential for the prevention of ETEC infections in pigs.
Collapse
|
11
|
Wu Z, Fan H, Jin J, Gao S, Huang R, Wu S, Bao W. Insight into mechanisms of pig lncRNA FUT3-AS1 regulating E. coli F18-bacterial diarrhea. PLoS Pathog 2022; 18:e1010584. [PMID: 35696408 PMCID: PMC9191744 DOI: 10.1371/journal.ppat.1010584] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 05/10/2022] [Indexed: 12/21/2022] Open
Abstract
Escherichia coli F18 is a common conditional pathogen that is associated with a variety of infections in humans and animals. LncRNAs have emerged as critical players in pathogen infection, but their role in the resistance of the host to bacterial diarrhea remains unknown. Here, we used piglets as animal model and identified an antisense lncRNA termed FUT3-AS1 as a host regulator related to E. coli F18 infection by RNA sequencing. Downregulation of FUT3-AS1 expression contributed to the enhancement of E. coli F18 resistance in IPEC-J2 cells. FUT3-AS1 knockdown reduced FUT3 expression via decreasing the H4K16ac level of FUT3 promoter. Besides, the FUT3-AS1/miR-212 axis could act as a competing endogenous RNA to regulate FUT3 expression. Functional analysis demonstrated that target FUT3 plays a vital role in the resistance of IPEC-J2 cells to E. coli F18 invasion. A Fut3-knockout mice model was established and Fut3-knockout mice obviously improved the ability of resistance to bacterial diarrhea. Interestingly, FUT3 could enhance E. coli F18 susceptibility by activating glycosphingolipid biosynthesis and toll-like receptor signaling which are related to receptor formation and immune response, respectively. In summary, we have identified a novel biomarker FUT3-AS1 that modulates E. coli F18 susceptibility via histone H4 modifications or miR-212/FUT3 axis, which will provide theoretical guidance to develop novel strategies for combating bacterial diarrhea in piglets.
Collapse
Affiliation(s)
- Zhengchang Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P. R. China
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, P. R. China
| | - Hairui Fan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P. R. China
| | - Jian Jin
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, P. R. China
| | - Song Gao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, P. R. China
| | - Ruihua Huang
- Institute of Swine Science, Nanjing Agricultural University, Nanjing, Jiangsu, P. R. China
| | - Shenglong Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P. R. China
| | - Wenbin Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, P. R. China
- * E-mail:
| |
Collapse
|
12
|
Zalem D, Juhás M, Terrinoni M, King-Lyons N, Lebens M, Varrot A, Connell TD, Teneberg S. Characterization of the ganglioside recognition profile of Escherichia coli heat-labile enterotoxin LT-IIc. Glycobiology 2022; 32:391-403. [PMID: 34972864 PMCID: PMC9022906 DOI: 10.1093/glycob/cwab133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/23/2021] [Accepted: 12/19/2021] [Indexed: 11/14/2022] Open
Abstract
The heat-labile enterotoxins of Escherichia coli and cholera toxin of Vibrio cholerae are related in structure and function. Each of these oligomeric toxins is comprised of one A polypeptide and five B polypeptides. The B-subunits bind to gangliosides, which are followed by uptake into the intoxicated cell and activation of the host's adenylate cyclase by the A-subunits. There are two antigenically distinct groups of these toxins. Group I includes cholera toxin and type I heat-labile enterotoxin of E. coli; group II contains the type II heat-labile enterotoxins of E. coli. Three variants of type II toxins, designated LT-IIa, LT-IIb and LT-IIc have been described. Earlier studies revealed the crystalline structure of LT-IIb. Herein the carbohydrate binding specificity of LT-IIc B-subunits was investigated by glycosphingolipid binding studies on thin-layer chromatograms and in microtiter wells. Binding studies using a large variety of glycosphingolipids showed that LT-IIc binds with high affinity to gangliosides with a terminal Neu5Acα3Gal or Neu5Gcα3Gal, e.g. the gangliosides GM3, GD1a and Neu5Acα3-/Neu5Gcα3--neolactotetraosylceramide and Neu5Acα3-/Neu5Gcα3-neolactohexaosylceramide. The crystal structure of LT-IIc B-subunits alone and with bound LSTd/sialyl-lacto-N-neotetraose d pentasaccharide uncovered the molecular basis of the ganglioside recognition. These studies revealed common and unique functional structures of the type II family of heat-labile enterotoxins.
Collapse
Affiliation(s)
- Dani Zalem
- Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, Institute of Biomedicine, University of Gothenburg, Sweden
| | - Martin Juhás
- Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, Institute of Biomedicine, University of Gothenburg, Sweden
- Department of Pharmaceutical Chemistry and Pharmaceutical Analysis, Charles University, Faculty of Pharmacy in Hradec Králové, Akademika Heyrovského 1203, Hradec Králové 500 05, Czech Republic
| | - Manuela Terrinoni
- Department of Microbiology and Immunology, Sahlgrenska Academy, Institute of Biomedicine, University of Gothenburg, Sweden
| | - Natalie King-Lyons
- Department of Microbiology & Immunology and The Witebsky Center for Microbial Pathogenesis and Immunology, The Jacobs School of Medicine and Biomedical Sciences, The University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Michael Lebens
- Department of Microbiology and Immunology, Sahlgrenska Academy, Institute of Biomedicine, University of Gothenburg, Sweden
| | | | - Terry D Connell
- Department of Microbiology & Immunology and The Witebsky Center for Microbial Pathogenesis and Immunology, The Jacobs School of Medicine and Biomedical Sciences, The University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Susann Teneberg
- Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, Institute of Biomedicine, University of Gothenburg, Sweden
| |
Collapse
|
13
|
Kim K, He Y, Jinno C, Kovanda L, Li X, Bravo D, Cox E, Liu Y. Supplementation of oligosaccharide-based polymer enhanced growth and disease resistance of weaned pigs by modulating intestinal integrity and systemic immunity. J Anim Sci Biotechnol 2022; 13:10. [PMID: 35016715 PMCID: PMC8753815 DOI: 10.1186/s40104-021-00655-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 11/21/2021] [Indexed: 11/18/2022] Open
Abstract
Background There is a great demand for antibiotic alternatives to maintain animal health and productivity. The objective of this experiment was to determine the efficacy of dietary supplementation of a blood group A6 type 1 antigen oligosaccharides-based polymer (Coligo) on growth performance, diarrhea severity, intestinal health, and systemic immunity of weaned pigs experimentally infected with an enterotoxigenic Escherichia coli (ETEC), when compared with antibiotics. Results Pigs in antibiotic carbadox or Coligo treatment groups had greater (P < 0.05) body weight on d 5 or d 11 post-inoculation (PI) than pigs in the control group, respectively. Supplementation of antibiotics or Coligo enhanced (P < 0.05) feed efficiency from d 0 to 5 PI and reduced (P < 0.05) frequency of diarrhea throughout the experiment, compared with pigs in the control group. Supplementation of antibiotics reduced (P < 0.05) fecal β-hemolytic coliforms on d 2, 5, and 8 PI. Pigs in antibiotics or Coligo groups had reduced (P < 0.05) neutrophil counts and serum haptoglobin concentration compared to pigs in the control group on d 2 and 5 PI. Pigs in Coligo had reduced (P < 0.05) total coliforms in mesenteric lymph nodes on d 5 and 11 PI, whereas pigs in antibiotics or Coligo groups had reduced (P < 0.05) total coliforms in spleen on d 11 PI compared with pigs in the control group. On d 5 PI, pigs in the Coligo group had greater (P < 0.05) gene expression of ZO1 in jejunal mucosa, but less (P < 0.05) mRNA expression of IL1B, IL6, and TNF in ileal mucosa, in comparison with pigs in the control group. Supplementation of antibiotics enhanced (P < 0.05) the gene expression of OCLN in jejunal mucosa but decreased (P < 0.05) IL1B and IL6 gene expression in ileal mucosa, compared with the control. On d 11 PI, supplementation of antibiotics or Coligo up-regulated (P < 0.05) gene expression of CLDN1 in jejunal mucosa, but Coligo reduced (P < 0.05) IL6 gene expression in ileal mucosa compared to pigs in the control group. Conclusions Supplementation of Coligo improved growth performance, alleviated diarrhea severity, and enhanced gut health in weaned pigs infected with ETEC F18 in a manner similar to in-feed antibiotics. Supplementary Information The online version contains supplementary material available at 10.1186/s40104-021-00655-2.
Collapse
Affiliation(s)
- Kwangwook Kim
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Yijie He
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Cynthia Jinno
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Lauren Kovanda
- Department of Animal Science, University of California, Davis, CA, 95616, USA
| | - Xunde Li
- School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | | | - Eric Cox
- Department of Virology, Parasitology and Immunology, Ghent University, 9000, Ghent, Belgium
| | - Yanhong Liu
- Department of Animal Science, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
14
|
Acetone Ingestion Mimics a Fasting State to Improve Glucose Tolerance in a Mouse Model of Gestational Hyperglycemia. Int J Mol Sci 2021; 22:ijms222312914. [PMID: 34884717 PMCID: PMC8657850 DOI: 10.3390/ijms222312914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/23/2021] [Accepted: 11/27/2021] [Indexed: 11/16/2022] Open
Abstract
Gestational diabetes mellitus results, in part, from a sub-optimal β-cell mass (BCM) during pregnancy. Artemisinins were reported to increase BCM in models of diabetes by α- to β-cell conversion leading to enhanced glucose tolerance. We used a mouse model of gestational glucose intolerance to compare the effects of an artemisinin (artesunate) on glycemia of pregnant mice with vehicle treatment (acetone) or no treatment. Animals were treated daily from gestational days (GD) 0.5 to 6.5. An intraperitoneal glucose tolerance test was performed prior to euthanasia at GD18.5 or post-partum. Glucose tolerance was significantly improved in both pregnant and non-pregnant mice with both artesunate and vehicle-alone treatment, suggesting the outcome was primarily due to the acetone vehicle. In non-pregnant, acetone-treated animals, improved glucose tolerance was associated with a higher BCM and a significant increase in bihormonal insulin and glucagon-containing pancreatic islet cells, suggesting α- to β-cell conversion. BCM did not differ with treatment during pregnancy or post-partum. However, placental weight was higher in acetone-treated animals and was associated with an upregulation of apelinergic genes. Acetone-treated animals had reduced weight gain during treatment despite comparable food consumption to non-treated mice, suggesting transient effects on nutrient uptake. The mean duodenal and ileum villus height was reduced following exposure to acetone. We conclude that acetone treatment may mimic transient fasting, resulting in a subsequent improvement in glucose tolerance during pregnancy.
Collapse
|
15
|
Van der Weken H, Sanz Garcia R, Sanders NN, Cox E, Devriendt B. Antibody-Mediated Targeting of Antigens to Intestinal Aminopeptidase N Elicits Gut IgA Responses in Pigs. Front Immunol 2021; 12:753371. [PMID: 34721427 PMCID: PMC8551371 DOI: 10.3389/fimmu.2021.753371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/21/2021] [Indexed: 12/03/2022] Open
Abstract
Many pathogens enter the host via the gut, causing disease in animals and humans. A robust intestinal immune response is necessary to protect the host from these gut pathogens. Despite being best suited for eliciting intestinal immunity, oral vaccination remains a challenge due to the gastrointestinal environment, a poor uptake of vaccine antigens by the intestinal epithelium and the tolerogenic environment pervading the gut. To improve uptake, efforts have focused on targeting antigens towards the gut mucosa. An interesting target is aminopeptidase N (APN), a conserved membrane protein present on small intestinal epithelial cells shown to mediate epithelial transcytosis. Here, we aimed to further optimize this oral vaccination strategy in a large animal model. Porcine APN-specific monoclonal antibodies were generated and the most promising candidate in terms of epithelial transcytosis was selected to generate antibody fusion constructs, comprising a murine IgG1 or porcine IgA backbone and a low immunogenic antigen: the F18-fimbriated E. coli tip adhesin FedF. Upon oral delivery of these recombinant antibodies in piglets, both mucosal and systemic immune responses were elicited. The presence of the FedF antigen however appeared to reduce these immune responses. Further analysis showed that F18 fimbriae were able to disrupt the antigen presenting capacity of intestinal antigen presenting cells, implying potential tolerogenic effects of FedF. Altogether, these findings show that targeted delivery of molecules to epithelial aminopeptidase N results in their transcytosis and delivery to the gut immune systems. The results provide a solid foundation for the development of oral subunit vaccines to protect against gut pathogens.
Collapse
Affiliation(s)
- Hans Van der Weken
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Raquel Sanz Garcia
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Niek N Sanders
- Laboratory of Gene therapy, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Eric Cox
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Bert Devriendt
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
16
|
DNA Methylation of Pig FUT3 Promoter Alters mRNA Expression to Regulate E. coli F18 Susceptibility. Genes (Basel) 2021; 12:genes12101586. [PMID: 34680980 PMCID: PMC8535959 DOI: 10.3390/genes12101586] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/30/2021] [Accepted: 10/02/2021] [Indexed: 01/15/2023] Open
Abstract
Post-weaning diarrhea (PWD) is frequently associated with E. coli F18 infections in piglets. However, the underlying molecular mechanism concerning the resistance of E. coli F18 in local weaned piglets in China is not clearly understood. In the present study, by a comparative analysis of the transcriptome, a-1,3-fucosyltransferase (FUT3) was evaluated as a key candidate correlated with resistance to E. coli F18 in Sutai and Meishan piglets. Functional analysis demonstrated that FUT3 acts as a key positive regulator of E. coli F18 susceptibility in newly food accustomed piglets. However, the core promoter of FUT3 was present at −500–(−206) bp (chr.2: g.73171117–g.73171616), comprising of 9 methylated CpG sites. Among these, the methylation levels of the two CpG sites (mC-3, mC-5) located in HIF1A and Sp1 transcription factor (TF) considerably associated with mRNA expression of FUT3 (p < 0.05). Our findings indicated that the methylation of mC-3 and mC-5 sites has certain inhibitory effect on FUT3 expression and promotes the resistance of E. coli F18 in piglets. The underlined study may explore FUT3 as a new candidate target in E. coli F18 infection in Chinese local weaned piglets.
Collapse
|
17
|
Exploiting pilus-mediated bacteria-host interactions for health benefits. Mol Aspects Med 2021; 81:100998. [PMID: 34294411 DOI: 10.1016/j.mam.2021.100998] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/30/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023]
Abstract
Surface pili (or fimbriae) are an important but conspicuous adaptation of several genera and species of Gram-negative and Gram-positive bacteria. These long and non-flagellar multi-subunit adhesins mediate the initial contact that a bacterium has with a host or environment, and thus have come to be regarded as a key colonization factor for virulence activity in pathogens or niche adaptation in commensals. Pili in pathogenic bacteria are well recognized for their roles in the adhesion to host cells, colonization of tissues, and establishment of infection. As an 'anti-adhesive' ploy, targeting pilus-mediated attachment for disruption has become a potentially effective alternative to using antibiotics. In this review, we give a description of the several structurally distinct bacterial pilus types thus far characterized, and as well offer details about the intricacy of their individual structure, assembly, and function. With a molecular understanding of pilus biogenesis and pilus-mediated host interactions also provided, we go on to describe some of the emerging new approaches and compounds that have been recently developed to prevent the adhesion, colonization, and infection of piliated bacterial pathogens.
Collapse
|
18
|
Vermeire B, Gonzalez LM, Jansens RJJ, Cox E, Devriendt B. Porcine small intestinal organoids as a model to explore ETEC-host interactions in the gut. Vet Res 2021; 52:94. [PMID: 34174960 PMCID: PMC8235647 DOI: 10.1186/s13567-021-00961-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/03/2021] [Indexed: 12/14/2022] Open
Abstract
Small intestinal organoids, or enteroids, represent a valuable model to study host–pathogen interactions at the intestinal epithelial surface. Much research has been done on murine and human enteroids, however only a handful studies evaluated the development of enteroids in other species. Porcine enteroid cultures have been described, but little is known about their functional responses to specific pathogens or their associated virulence factors. Here, we report that porcine enteroids respond in a similar manner as in vivo gut tissues to enterotoxins derived from enterotoxigenic Escherichia coli, an enteric pathogen causing postweaning diarrhoea in piglets. Upon enterotoxin stimulation, these enteroids not only display a dysregulated electrolyte and water balance as shown by their swelling, but also secrete inflammation markers. Porcine enteroids grown as a 2D-monolayer supported the adhesion of an F4+ ETEC strain. Hence, these enteroids closely mimic in vivo intestinal epithelial responses to gut pathogens and are a promising model to study host–pathogen interactions in the pig gut. Insights obtained with this model might accelerate the design of veterinary therapeutics aimed at improving gut health.
Collapse
Affiliation(s)
- Bjarne Vermeire
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Laboratory of Immunology, Ghent University, 9820, Merelbeke, Belgium
| | - Liara M Gonzalez
- Laboratory of Intestinal Regenerative Medicine, College of Veterinary Medicine, NCSU, Raleigh, NC, USA
| | - Robert J J Jansens
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Laboratory of Immunology, Ghent University, 9820, Merelbeke, Belgium
| | - Eric Cox
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Laboratory of Immunology, Ghent University, 9820, Merelbeke, Belgium
| | - Bert Devriendt
- Department of Virology, Parasitology, Immunology, Faculty of Veterinary Medicine, Laboratory of Immunology, Ghent University, 9820, Merelbeke, Belgium.
| |
Collapse
|
19
|
Galeev A, Suwandi A, Cepic A, Basu M, Baines JF, Grassl GA. The role of the blood group-related glycosyltransferases FUT2 and B4GALNT2 in susceptibility to infectious disease. Int J Med Microbiol 2021; 311:151487. [PMID: 33662872 DOI: 10.1016/j.ijmm.2021.151487] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/01/2021] [Accepted: 02/23/2021] [Indexed: 12/12/2022] Open
Abstract
The glycosylation profile of the gastrointestinal tract is an important factor mediating host-microbe interactions. Variation in these glycan structures is often mediated by blood group-related glycosyltransferases, and can lead to wide-ranging differences in susceptibility to both infectious- as well as chronic disease. In this review, we focus on the interplay between host glycosylation, the intestinal microbiota and susceptibility to gastrointestinal pathogens based on studies of two exemplary blood group-related glycosyltransferases that are conserved between mice and humans, namely FUT2 and B4GALNT2. We highlight that differences in susceptibility can arise due to both changes in direct interactions, such as bacterial adhesion, as well as indirect effects mediated by the intestinal microbiota. Although a large body of experimental work exists for direct interactions between host and pathogen, determining the more complex and variable mechanisms underlying three-way interactions involving the intestinal microbiota will be the subject of much-needed future research.
Collapse
Affiliation(s)
- Alibek Galeev
- Max Planck Institute for Evolutionary Biology, Plön, Germany and Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Abdulhadi Suwandi
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany
| | - Aleksa Cepic
- Max Planck Institute for Evolutionary Biology, Plön, Germany and Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Meghna Basu
- Max Planck Institute for Evolutionary Biology, Plön, Germany and Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - John F Baines
- Max Planck Institute for Evolutionary Biology, Plön, Germany and Institute for Experimental Medicine, Kiel University, Kiel, Germany.
| | - Guntram A Grassl
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School and German Center for Infection Research (DZIF), Hannover, Germany.
| |
Collapse
|
20
|
Von Mentzer A, Zalem D, Chrienova Z, Teneberg S. Colonization factor CS30 from enterotoxigenic Escherichia coli binds to sulfatide in human and porcine small intestine. Virulence 2021; 11:381-390. [PMID: 32245341 PMCID: PMC7161690 DOI: 10.1080/21505594.2020.1749497] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The ability to adhere via colonization factors to specific receptors located on the intestinal mucosa is a key virulence factor in enterotoxigenic Escherichia coli (ETEC) pathogenesis. Here, the potential glycosphingolipid receptors of the novel human ETEC colonization factor CS30 were examined by binding of CS30-expressing bacteria to glycosphingolipids on thin-layer chromatograms. We thereby found a highly specific binding of CS30-expressing bacteria to a fast-migrating acid glycosphingolipid of human and porcine small intestine, while no binding was obtained with a mutant ETEC strain unable to express CS30 fimbriae. The CS30 binding glycosphingolipid from human small intestine was isolated and characterized by mass spectrometry as sulfatide (SO3-3Galβ1Cer). Comparative binding studies using sulfatides with different ceramide compositions gave a preferential binding of CS30 to sulfatide with d18:1-h24:0 ceramide. This ceramide species of sulfatide was also isolated from human small intestine and characterized by mass spectrometry and antibody binding. These studies implicate sulfatide as candidate receptor for mediating attachment of CS30-fimbriated ETEC to human and porcine small intestinal cells. Our findings may be a basis for designing receptor saccharide analogues for inhibition of the intestinal adhesion of CS30-expressing E. coli.
Collapse
Affiliation(s)
- Astrid Von Mentzer
- Department of Microbiology and Immunology, Sahlgrenska Academy, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.,Wellcome Sanger Institute: Parasites and Microbes Programme, Hinxton, UK
| | - Dani Zalem
- Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Zofia Chrienova
- Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.,Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czech Republic
| | - Susann Teneberg
- Department of Medical Biochemistry and Cell Biology, Sahlgrenska Academy, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
21
|
Porcine and Bovine Forms of Lactoferrin Inhibit Growth of Porcine Enterotoxigenic Escherichia coli and Degrade Its Virulence Factors. Appl Environ Microbiol 2020; 86:AEM.00524-20. [PMID: 32631861 DOI: 10.1128/aem.00524-20] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/07/2020] [Indexed: 12/19/2022] Open
Abstract
Postweaning diarrhea (PWD) is an economically important, multifactorial disease affecting pigs within the first 2 weeks after weaning. The most common agent associated with PWD is enterotoxigenic Escherichia coli (ETEC). Currently, antibiotics are used to control PWD, and this has most likely contributed to an increased prevalence of antibiotic-resistant strains. This puts pressure on veterinarians and farmers to decrease or even abandon the use of antibiotics, but these measures need to be supported by alternative strategies for controlling these infections. Naturally derived molecules, such as lactoferrin, could be potential candidates due to their antibacterial or immune-modulating activities. Here, we analyzed the ability of bovine lactoferrin (bLF), porcine lactoferrin (pLF), and ovotransferrin (ovoTF) to inhibit ETEC growth, degrade ETEC virulence factors, and inhibit adherence of these pathogens to porcine intestinal epithelial cells. Our results revealed that bLF and pLF, but not ovoTF, inhibit the growth of ETEC. Furthermore, bLF and pLF can degrade several virulence factors produced by ETEC strains, more specifically F4 fimbriae, F18 fimbriae, and flagellin. On the other hand, ovoTF degrades F18 fimbriae and flagellin but not F4 fimbriae. An in vitro adhesion assay showed that bLF, ovoTF, and pLF can decrease the number of bacteria adherent to epithelial cells. Our findings demonstrate that lactoferrin can directly affect porcine ETEC strains, which could allow lactoferrin to serve as an alternative to antimicrobials for the prevention of ETEC infections in piglets.IMPORTANCE Currently, postweaning F4+ and F18+ Escherichia coli infections in piglets are controlled by the use of antibiotics and zinc oxide, but the use of these antimicrobial agents most likely contributes to an increase in antibiotic resistance. Our work demonstrates that bovine and porcine lactoferrin can inhibit the growth of porcine enterotoxigenic E. coli strains. In addition, we also show that lactoferrin can reduce the adherence of these strains to small intestinal epithelial cells, even at a concentration that does not inhibit bacterial growth. This research could allow us to develop lactoferrin as an alternative strategy to prevent enterotoxigenic E. coli (ETEC) infections in piglets.
Collapse
|
22
|
The Intriguing Interaction of Escherichia coli with the Host Environment and Innovative Strategies To Interfere with Colonization: a Summary of the 2019 E. coli and the Mucosal Immune System Meeting. Appl Environ Microbiol 2020; 86:AEM.02085-20. [PMID: 33008822 DOI: 10.1128/aem.02085-20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The third E. coli and the Mucosal Immune System (ECMIS) meeting was held at Ghent University in Belgium from 2 to 5 June 2019. It brought together an international group of scientists interested in mechanisms of colonization, host response, and vaccine development. ECMIS distinguishes itself from related meetings on these enteropathogens by providing a greater emphasis on animal health and disease and covering a broad range of pathotypes, including enterohemorrhagic, enteropathogenic, enterotoxigenic, enteroaggregative, and extraintestinal pathogenic Escherichia coli As it is well established that the genus Shigella represents a subspecies of E. coli, these organisms along with related enteroinvasive E. coli are also included. In addition, Tannerella forsythia, a periodontal pathogen, was presented as an example of a pathogen which uses its surface glycans for mucosal interaction. This review summarizes several highlights from the 2019 meeting and major advances to our understanding of the biology of these pathogens and their impact on the host.
Collapse
|
23
|
Josenhans C, Müthing J, Elling L, Bartfeld S, Schmidt H. How bacterial pathogens of the gastrointestinal tract use the mucosal glyco-code to harness mucus and microbiota: New ways to study an ancient bag of tricks. Int J Med Microbiol 2020; 310:151392. [DOI: 10.1016/j.ijmm.2020.151392] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/28/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022] Open
|
24
|
Mottram L, Chakraborty S, Cox E, Fleckenstein J. How genomics can be used to understand host susceptibility to enteric infection, aiding in the development of vaccines and immunotherapeutic interventions. Vaccine 2019; 37:4805-4810. [PMID: 30709726 PMCID: PMC6663652 DOI: 10.1016/j.vaccine.2019.01.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/26/2018] [Accepted: 01/10/2019] [Indexed: 12/27/2022]
Abstract
Thanks to the modern sequencing era, the extent to which infectious disease imposes selective pressures on the worldwide human population is being revealed. This is aiding our understanding of the underlying immunological and host mechanistic defenses against these pathogens, as well as potentially assisting in the development of vaccines and therapeutics to control them. As a consequence, the workshop "How genomics can be used to understand host susceptibility to enteric infection, aiding in the development of vaccines and immunotherapeutic interventions" at the VASE 2018 meeting, aimed to discuss how genomics and related tools could be used to assist Shigella and ETEC vaccine development. The workshop featured four short presentations which highlighted how genomic applications can be used to assist in the identification of genetic patterns related to the virulence of disease, or host genetic factors that could contribute to immunity or successful vaccine responses. Following the presentations, there was an open debate with workshop attendees to discuss the best ways to utilise such genomic studies, to improve or accelerate the process of both Shigella and ETEC vaccine development. The workshop concluded by making specific recommendations on how genomic research methods could be strengthened and harmonised within the ETEC and Shigella research communities.
Collapse
Affiliation(s)
- Lynda Mottram
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| | - Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Eric Cox
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - James Fleckenstein
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States; Medicine Service, Veterans Affairs Medical Center, St. Louis, MO, United States
| |
Collapse
|
25
|
Mani S, Toapanta FR, McArthur MA, Qadri F, Svennerholm AM, Devriendt B, Phalipon A, Cohen D, Sztein MB. Role of antigen specific T and B cells in systemic and mucosal immune responses in ETEC and Shigella infections, and their potential to serve as correlates of protection in vaccine development. Vaccine 2019; 37:4787-4793. [PMID: 31230883 PMCID: PMC7413037 DOI: 10.1016/j.vaccine.2019.03.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/07/2019] [Accepted: 03/19/2019] [Indexed: 12/13/2022]
Abstract
The generation of robust systemic and mucosal antibody and cell-mediated immune (CMI) responses that are protective, long-lasting, and can quickly be recalled upon subsequent re-exposure to the cognate antigen is the key to the development of effective vaccine candidates. These responses, whether they represent mechanistic or non-mechanistic immunological correlates of protection, usually entail the activation of T cell memory and effector subsets (T-CMI) and induction of long-lasting memory B cells. However, for ETEC and Shigella, the precise role of these key immune cells in primary and secondary (anamnestic) immune responses remains ill-defined. A workshop to address immune correlates for ETEC and Shigella, in general, and to elucidate the mechanistic role of T-cell subsets and B-cells, both systemically and in the mucosal microenvironment, in the development of durable protective immunity against ETEC and Shigella was held at the recent 2nd Vaccines against Shigella and ETEC (VASE) conference in June 2018. This report is a summary of the presentations and the discussion that ensued at the workshop.
Collapse
Affiliation(s)
| | - Franklin R Toapanta
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Monica A McArthur
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Firdausi Qadri
- Infectious Diseases Division, International Center for Diarrheal Diseases Research, Dhaka, Bangladesh
| | - Ann-Mari Svennerholm
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Sweden
| | - Bert Devriendt
- Faculty of Veterinary Medicine, Department of Virology, Parasitology, and Immunology, Ghent University, Belgium
| | - Armelle Phalipon
- Molecular Microbial Pathogenesis, INSERM U1202, Institut Pasteur, Paris, France
| | - Daniel Cohen
- School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
26
|
Resistance to ETEC F4/F18-mediated piglet diarrhoea: opening the gene black box. Trop Anim Health Prod 2019; 51:1307-1320. [PMID: 31127494 DOI: 10.1007/s11250-019-01934-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 05/15/2019] [Indexed: 01/08/2023]
Abstract
Diarrhoea, a significant problem in pig rearing industry affecting pre- and post-weaning piglets is caused by enterotoxigenic Escherichia coli (ETEC). The ETEC are classified as per the fimbriae types which are responsible for bacterial attachment with enterocytes and release of toxins causing diarrhoea. However, genetic difference exists for susceptibility to ETEC infection in piglets. The different phenotypes found in pigs determine their (pigs') susceptibility or resistance towards fimbrial subtypes/variants (F4ab, F4ac, F4ad and F18). Specific receptors are present on intestinal epithelium for attachment of these fimbriae, which do not express to same level in all animals. This differential expression is genetically determined and thus their genetic causes (may be putative candidate gene or mutations) render some animals resistant or susceptible to one or more fimbrial subtypes. Genetic linkage studies have revealed the mapping location of the receptor loci for the two most frequent variants F4ab and F4ac to SSC13q41 (i.e. q arm of 13th chromosome of Sus scrofa). Some SNPs have been identified in mucin gene family, transferring receptor gene, fucosyltransferase 1 gene and swine leucocyte antigen locus that are proposed to be linked mutations for resistance/susceptibility towards ETEC diarrhoea. However, owing to the variety of fimbrial types and subtypes, it would be difficult to identify a single causative mutation and the candidate loci may involve more number of genes/regions. In this review, we focus on the genetic mutations in genes involved in imparting resistance/susceptibility to F4 or F18 ETEC diarrhoea and possibilities to use them as marker for selection against susceptible animals.
Collapse
|
27
|
Motta V, Luise D, Bosi P, Trevisi P. Faecal microbiota shift during weaning transition in piglets and evaluation of AO blood types as shaping factor for the bacterial community profile. PLoS One 2019; 14:e0217001. [PMID: 31095619 PMCID: PMC6522051 DOI: 10.1371/journal.pone.0217001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 05/02/2019] [Indexed: 12/31/2022] Open
Abstract
The host-microbiota interplay is recognized as a key factor for the homeostatic maintenance in animals. In pigs, the weaning transition represents a drastic changes event leading to high risk of gut dysbiosis, which in most cases results in economic losses for swine industry. The blood type antigens expressed on mucosal surfaces can act as receptors for bacterial adhesion and the hypothesis of possible associations between blood groups and intestinal microbial profiles has been tested in human with contrasting results. Nevertheless, no studies testing the blood type as possible shaping factor for gut microbiota are available for pigs. The results of our previous study suggested the porcine AO blood types system as a possible factor influencing the microbiota composition. In the present study, the changes in fecal microbiota of 12 piglets were followed from 7 days after birth to 2 weeks post-weaning, testing the hypothesis that blood types may impact on its structure. No effects attributable to the difference in blood groups were detected, however, the sampling site (faeces) and the low statistical power might have masked the hypothesized impact. The data clearly showed the rearrangement of the bacterial ecosystem triggered by weaning transition; mainly consisting of a shift from a Bacteroidaceae-Enterobacteriaceae dominated community, to a Prevotellaceae-Ruminococcaceae dominated community. The functional analysis by metagenomic predictions suggested a role of the high levels of long-chain fatty acid in swine milk as energy source for Enterobacteriaceae (E. coli), in suckling piglets. This study provides a first insight for further investigations; indicating the need for larger sample size, preferably derived from intestinal mucosa, to test the potential effect of blood groups on gut microbiota profiles, and for analyses aimed at assessing the long-chain fatty acids degradation activity within the intestinal microbiota of suckling piglets, with particular attention to the role of E. coli.
Collapse
Affiliation(s)
- Vincenzo Motta
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Diana Luise
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Paolo Bosi
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Paolo Trevisi
- Department of Agricultural and Food Sciences (DISTAL), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| |
Collapse
|
28
|
Wu Z, Feng H, Cao Y, Huang Y, Dai C, Wu S, Bao W. New Insight into the Molecular Mechanism of the FUT2 Regulating Escherichia coli F18 Resistance in Weaned Piglets. Int J Mol Sci 2018; 19:E3301. [PMID: 30352970 PMCID: PMC6275016 DOI: 10.3390/ijms19113301] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 10/10/2018] [Indexed: 11/23/2022] Open
Abstract
Escherichia coli (E. coli) F18 is the main pathogen responsible for post-weaning diarrhea (PWD) in piglets. Resistance to E. coli F18 depends on the expression of the cognate receptors in the intestinal epithelial cells. However, the molecular mechanism of E. coli F18 resistance in weaned piglets remains unclear. Here, we performed a comparative transcriptome study of the duodenal tissue from Sutai E. coli F18 sensitive and resistant pigs by RNA-seq, and pig α(1,2) fucosyltransferase 2 (FUT2) was identified as a host differentially expressed gene controlling the E. coli F18 infection. Function analysis showed that the FUT2 expression was high in the duodenum and jejunum, with higher levels detected in sensitive individuals than in resistant individuals (p < 0.01). Expression levels of FUT2 were upregulated in IPEC-J2 cells after lipopolysaccharide (LPS)-induction or E. coli stimulation. FUT2 knockdown decreased the adhesion of E. coli F18 to IPEC-J2 cells (p < 0.05). FUT2 overexpression markedly increased the adhesion of E. coli F18 to IPEC-J2 cells (p < 0.05 or p < 0.01). Furthermore, the FUT2 mRNA levels correlated with methylation levels of the mC-22 site in the specificity protein 1 (Sp1) transcription factor (p < 0.05). Electrophoretic mobility shift assays (EMSA) showed that Sp1 interacts with the wild-type FUT2 promoter DNA, but not with methylated DNA. Our data suggested that FUT2 methylation at the mC-22 site inhibits Sp1 binding to the FUT2 promoter, thereby reducing FUT2 expression and enhancing E. coli F18 resistance in weaned piglets. These observations highlight FUT2 as a promising new target for combating E. coli F18 susceptibility in weaned piglets.
Collapse
Affiliation(s)
- Zhengchang Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Haiyue Feng
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Yue Cao
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Yanjie Huang
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Chaohui Dai
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| | - Shenglong Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou 225009, China.
| | - Wenbin Bao
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
29
|
Glyco-engineered cell line and computational docking studies reveals enterotoxigenic Escherichia coli CFA/I fimbriae bind to Lewis a glycans. Sci Rep 2018; 8:11250. [PMID: 30050155 PMCID: PMC6062558 DOI: 10.1038/s41598-018-29258-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 07/09/2018] [Indexed: 12/16/2022] Open
Abstract
We have previously reported clinical data to suggest that colonization factor I (CFA/I) fimbriae of enterotoxigenic Escherichia coli (ETEC) can bind to Lewis a (Lea), a glycan epitope ubiquitous in the small intestinal mucosa of young children (<2 years of age), and individuals with a genetic mutation of FUT2. To further elucidate the physiological binding properties of this interaction, we engineered Chinese Hamster Ovary (CHO-K1) cells to express Lea or Leb determinants on both N- and O-glycans. We used our glyco-engineered CHO-K1 cell lines to demonstrate that CfaB, the major subunit of ETEC CFA/I fimbriae, as well as four related ETEC fimbriae, bind more to our CHO-K1 cell-line expressing Lea, compared to cells carrying Leb or the CHO-K1 wild-type glycan phenotype. Furthermore, using in-silico docking analysis, we predict up to three amino acids (Glu25, Asn27, Thr29) found in the immunoglobulin (Ig)-like groove region of CfaB of CFA/I and related fimbriae, could be important for the preferential and higher affinity binding of CFA/I fimbriae to the potentially structurally flexible Lea glycan. These findings may lead to a better molecular understanding of ETEC pathogenesis, aiding in the development of vaccines and/or anti-infection therapeutics.
Collapse
|
30
|
Monedero V, Buesa J, Rodríguez-Díaz J. The Interactions between Host Glycobiology, Bacterial Microbiota, and Viruses in the Gut. Viruses 2018; 10:v10020096. [PMID: 29495275 PMCID: PMC5850403 DOI: 10.3390/v10020096] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 02/20/2018] [Accepted: 02/22/2018] [Indexed: 12/11/2022] Open
Abstract
Rotavirus (RV) and norovirus (NoV) are the major etiological agents of viral acute gastroenteritis worldwide. Host genetic factors, the histo-blood group antigens (HBGA), are associated with RV and NoV susceptibility and recent findings additionally point to HBGA as a factor modulating the intestinal microbial composition. In vitro and in vivo experiments in animal models established that the microbiota enhances RV and NoV infection, uncovering a triangular interplay between RV and NoV, host glycobiology, and the intestinal microbiota that ultimately influences viral infectivity. Studies on the microbiota composition in individuals displaying different RV and NoV susceptibilities allowed the identification of potential bacterial biomarkers, although mechanistic data on the virus-host-microbiota relation are still needed. The identification of the bacterial and HBGA interactions that are exploited by RV and NoV would place the intestinal microbiota as a new target for alternative therapies aimed at preventing and treating viral gastroenteritis.
Collapse
Affiliation(s)
- Vicente Monedero
- Department of Food Biotechnology, Institute of Agrochemistry and Food Technology (IATA, CSIC), Av Catedrático Agustín Escardino, 7, 46980 Paterna, Spain.
| | - Javier Buesa
- Departament of Microbiology, Faculty of Medicine, University of Valencia, Av. Blasco Ibañez 17, 46010 Valencia, Spain.
| | - Jesús Rodríguez-Díaz
- Departament of Microbiology, Faculty of Medicine, University of Valencia, Av. Blasco Ibañez 17, 46010 Valencia, Spain.
| |
Collapse
|
31
|
Lawan A, Jesse FFA, Idris UH, Odhah MN, Arsalan M, Muhammad NA, Bhutto KR, Peter ID, Abraham GA, Wahid AH, Mohd-Azmi ML, Zamri-Saad M. Mucosal and systemic responses of immunogenic vaccines candidates against enteric Escherichia coli infections in ruminants: A review. Microb Pathog 2018; 117:175-183. [PMID: 29471137 DOI: 10.1016/j.micpath.2018.02.039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 02/17/2018] [Accepted: 02/18/2018] [Indexed: 02/06/2023]
Abstract
Innumerable Escherichia coli of animal origin are identified, which are of economic significance, likewise, cattle, sheep and goats are the carrier of enterohaemorrhagic E. coli, which are less pathogenic, and can spread to people by way of direct contact and through the contamination of foodstuff or portable drinking water, causing serious illness. The immunization of ruminants has been carried out for ages and is largely acknowledged as the most economical and maintainable process of monitoring E. coli infection in ruminants. Yet, only a limited number of E. coli vaccines are obtainable. Mucosal surfaces are the most important ingress for E. coli and thus mucosal immune responses function as the primary means of fortification. Largely contemporary vaccination processes are done by parenteral administration and merely limited number of E. coli vaccines are inoculated via mucosal itinerary, due to its decreased efficacy. Nevertheless, aiming at maximal mucosal partitions to stimulate defensive immunity at both mucosal compartments and systemic site epitomises a prodigious task. Enormous determinations are involved in order to improve on novel mucosal E. coli vaccines candidate by choosing apposite antigens with potent immunogenicity, manipulating novel mucosal itineraries of inoculation and choosing immune-inducing adjuvants. The target of E. coli mucosal vaccines is to stimulate a comprehensive, effective and defensive immunity by specifically counteracting the antibodies at mucosal linings and by the stimulation of cellular immunity. Furthermore, effective E. coli mucosal vaccine would make vaccination measures stress-free and appropriate for large number of inoculation. On account of contemporary advancement in proteomics, metagenomics, metabolomics and transcriptomics research, a comprehensive appraisal of the immeasurable genes and proteins that were divulged by a bacterium is now in easy reach. Moreover, there exist marvellous prospects in this bourgeoning technologies in comprehending the host bacteria affiliation. Accordingly, the flourishing knowledge could massively guarantee to the progression of immunogenic vaccines against E. coli infections in both humans and animals. This review highlight and expounds on the current prominence of mucosal and systemic immunogenic vaccines for the prevention of E. coli infections in ruminants.
Collapse
Affiliation(s)
- A Lawan
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Department of Veterinary Medicine, Faculty of Veterinary Medicine, University of Maiduguri, Nigeria.
| | - F F A Jesse
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Department of Farm & Exotic Animals Medicine & Surgery, Faculty of Veterinary Medicine, Universiti Putra Malaysia (UPM), 43400 UPM, Serdang, Selangor, Malaysia
| | - U H Idris
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Department of Veterinary Public Health and Preventive Medicine, Faculty of Veterinary Medicine, University of Maiduguri, Nigeria
| | - M N Odhah
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Department of Veterinary Medicine, Faculty of Agriculture and Veterinary Medicine, Thamar University, Yemen
| | - M Arsalan
- Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, Malaysia; Livestock and Dairy Development Department Baluchistan, Pakistan
| | - N A Muhammad
- Institute of Tropical Agriculture and Food Security, Universiti Putra Malaysia, Malaysia
| | - K R Bhutto
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Veterinary Research & Diagnosis, Livestock and Fisheries Department, Sindh, Pakistan
| | - I D Peter
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Department of Theriogenology, Faculty of Veterinary Medicine, University of Maiduguri, Nigeria
| | - G A Abraham
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia; Department of Farm & Exotic Animals Medicine & Surgery, Faculty of Veterinary Medicine, Universiti Putra Malaysia (UPM), 43400 UPM, Serdang, Selangor, Malaysia
| | - A H Wahid
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - M L Mohd-Azmi
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Malaysia
| | - M Zamri-Saad
- Department of Veterinary Pathology and Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, Malaysia
| |
Collapse
|
32
|
Wang H, Wu S, Wu J, Sun S, Wu S, Bao W. Association analysis of the SNP (rs345476947) in the FUT2 gene with the production and reproductive traits in pigs. Genes Genomics 2018; 40:199-206. [PMID: 29892924 DOI: 10.1007/s13258-017-0623-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/15/2017] [Indexed: 12/21/2022]
Abstract
The FUT2 gene was considered as an important candidate for pathogenic infections, while the potential associations between this gene and the production and reproductive traits of pigs have not been explored. In this study, we detected the genetic variants of porcine FUT2 gene and analyzed the associations of the polymorphisms with FUT2 mRNA expression and production and reproductive traits (age at 100 kg, backfat thickness at 100 kg, eye muscle thickness, the number of newborn piglets, the number of weaned piglets, and birth weight) in 100 Large White sows. One single nucleotide polymorphism (SNP) (rs345476947, C→T) in the intron of FUT2 and three genotypes (TT, CT and CC) were determined. Association analysis revealed significant associations between this SNP with the number of newborn piglets and weaned piglets. Furthermore, individuals with the TT genotype had significantly higher numbers of newborn piglets and weaned piglets than those with the CC genotype (P < 0.05). Quantitative PCR analysis showed that FUT2 expression in individuals with CC genotype was significantly higher than those with TT and CT genotypes in the liver and lymph gland (P < 0.05) and higher than that of CT in the spleen, kidney, and duodenum (P < 0.05). These findings indicated that the TT genotype may be a favorable genotype for the reproductive traits of pigs. Our study revealed the genetic variants of the FUT2 gene and identified a promising candidate SNP (rs345476947) associated with the reproductive traits, which has the potential to be applied in selective breeding of pigs.
Collapse
Affiliation(s)
- Haifei Wang
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Sen Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Jiayun Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Shouyong Sun
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Shenglong Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, People's Republic of China
- Joint International Research Laboratory of Agriculture & Agri-Product SafetyYangzhou University, Yangzhou, 225009, People's Republic of China
| | - Wenbin Bao
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, People's Republic of China.
- Joint International Research Laboratory of Agriculture & Agri-Product SafetyYangzhou University, Yangzhou, 225009, People's Republic of China.
| |
Collapse
|
33
|
Coddens A, Loos M, Vanrompay D, Remon JP, Cox E. Cranberry extract inhibits in vitro adhesion of F4 and F18 +Escherichia coli to pig intestinal epithelium and reduces in vivo excretion of pigs orally challenged with F18 + verotoxigenic E. coli. Vet Microbiol 2017; 202:64-71. [PMID: 28161211 DOI: 10.1016/j.vetmic.2017.01.019] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 01/11/2017] [Accepted: 01/17/2017] [Indexed: 12/25/2022]
Abstract
F4+E. coli and F18+E. coli infections are an important threat for pig industry worldwide. Antibiotics are commonly used to treat infected piglets, but the emerging development of resistance against antibiotics raises major concerns. Hence, alternative therapies to prevent pigs from F4+E. coli and F18+E. coli infections need to be developed. Since cranberry previously showed anti-adhesive activity against uropathogenic E. coli, we aimed to investigate whether cranberry extract could also inhibit binding of F4+E. coli and F18+E. coli to pig intestinal epithelium. Using the in vitro villus adhesion assay, we found that low concentrations of cranberry extract (20μg or 100μg/ml) have strong inhibitory activity on F4+E. coli (75.3%, S.D.=9.31 or 95.8%, S.D.=2.56, respectively) and F18+E. coli adherence (100% inhibition). This effect was not due to antimicrobial activity. Moreover, cranberry extract (10mg or 100mg) could also abolish in vivo binding of F4 and F18 fimbriae to the pig intestinal epithelium in ligated loop experiments. Finally, two challenge experiments with F18+E. coli were performed to address the efficacy of in-feed or water supplemented cranberry extract. No effect could be observed in piglets that received cranberry extract only in feed (1g/kg or 10g/kg). However, supplementation of feed (10g/kg) and drinking water (1g/L) significantly decreased excretion and diarrhea. The decreased infection resulted in a decreased serum antibody response indicating reduced exposure to F18+E. coli.
Collapse
Affiliation(s)
- Annelies Coddens
- Laboratory of Veterinary Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium.
| | - Michaela Loos
- Laboratory of Veterinary Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium.
| | - Daisy Vanrompay
- Laboratory for Immunology and Animal Biotechnology, Department of Animal Production, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium.
| | - Jean Paul Remon
- Laboratory of Pharmaceutical Technology, Ghent University, Harelbekestraat 72, 9000 Gent, Belgium.
| | - Eric Cox
- Laboratory of Veterinary Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium.
| |
Collapse
|
34
|
Fecal Microbiota and Metabolome in a Mouse Model of Spontaneous Chronic Colitis: Relevance to Human Inflammatory Bowel Disease. Inflamm Bowel Dis 2016; 22:2767-2787. [PMID: 27824648 DOI: 10.1097/mib.0000000000000970] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Dysbiosis of the gut microbiota may be involved in the pathogenesis of inflammatory bowel disease (IBD). However, the mechanisms underlying the role of the intestinal microbiome and metabolome in IBD onset and its alteration during active treatment and recovery remain unknown. Animal models of chronic intestinal inflammation with similar microbial and metabolomic profiles would enable investigation of these mechanisms and development of more effective treatments. Recently, the Winnie mouse model of colitis closely representing the clinical symptoms and characteristics of human IBD has been developed. In this study, we have analyzed fecal microbial and metabolomic profiles in Winnie mice and discussed their relevance to human IBD. METHODS The 16S rRNA gene was sequenced from fecal DNA of Winnie and C57BL/6 mice to define operational taxonomic units at ≥97% similarity threshold. Metabolomic profiling of the same fecal samples was performed by gas chromatography-mass spectrometry. RESULTS Composition of the dominant microbiota was disturbed, and prominent differences were evident at all levels of the intestinal microbiome in fecal samples from Winnie mice, similar to observations in patients with IBD. Metabolomic profiling revealed that chronic colitis in Winnie mice upregulated production of metabolites and altered several metabolic pathways, mostly affecting amino acid synthesis and breakdown of monosaccharides to short chain fatty acids. CONCLUSIONS Significant dysbiosis in the Winnie mouse gut replicates many changes observed in patients with IBD. These results provide justification for the suitability of this model to investigate mechanisms underlying the role of intestinal microbiota and metabolome in the pathophysiology of IBD.
Collapse
|
35
|
Wu Z, Qin W, Wu S, Zhu G, Bao W, Wu S. Identification of microRNAs regulating Escherichia coli F18 infection in Meishan weaned piglets. Biol Direct 2016; 11:59. [PMID: 27809935 PMCID: PMC5093996 DOI: 10.1186/s13062-016-0160-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/20/2016] [Indexed: 12/21/2022] Open
Abstract
Background Escherichia coli F18 is mainly responsible for post-weaning diarrhea (PWD) in piglets. The molecular regulation of E. coli F18 resistance in Chinese domestic weaned piglets is still obscure. We used Meishan piglets as model animals to test their susceptibility to E. coli F18. Small RNA duodenal libraries were constructed for E. coli F18-sensitive and -resistant weaned piglets challenged with E. coli F18 and sequenced using Illumina Solexa high-throughput sequencing technology. Results Sequencing results showed that 3,475,231 and 37,198,259 clean reads were obtained, with 311 known miRNAs differently expressed in resistant and sensitive groups, respectively. Twenty-four miRNAs, including 15 up-regulated and 9 down-regulated, demonstrated more than a 2-fold differential expression between the F18-resistant and -sensitive piglets. Stem-loop RT-qPCR showed that miR-136, miR-196b, miR-499-5p and miR-218-3p significantly expressed in intestinal tissue (p < 0.05). KEGG pathway analysis for target genes revealed that differently expressed miRNAs were involved in infectious diseases, signal transduction and immune system pathways. Interestingly, the expression of miR-218-3p in intestinal tissue had a very significant negative correlation with target DLG5 (P < 0.01). Conclusions Based on the expression correlation between miRNA and target genes analysis, we speculate that miR-218-3p targeting to DLG5, appears to be very promising candidate for miRNAs involved in response to E. coli F18 infection. The present study provides improved database information on pig miRNAs, better understanding of the genetic basis of E. coli F18 resistance in local Chinese pig breeds and lays a new foundation for identifying novel markers of E. coli F18 resistance. Reviewers This article was reviewed by Neil R Smalheiser and Weixiong Zhang. Electronic supplementary material The online version of this article (doi:10.1186/s13062-016-0160-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhengchang Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Weiyun Qin
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Seng Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, People's Republic of China
| | - Wenbin Bao
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Shenglong Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, People's Republic of China.
| |
Collapse
|
36
|
Dubreuil JD, Isaacson RE, Schifferli DM. Animal Enterotoxigenic Escherichia coli. EcoSal Plus 2016; 7:10.1128/ecosalplus.ESP-0006-2016. [PMID: 27735786 PMCID: PMC5123703 DOI: 10.1128/ecosalplus.esp-0006-2016] [Citation(s) in RCA: 193] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Indexed: 12/13/2022]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is the most common cause of E. coli diarrhea in farm animals. ETEC are characterized by the ability to produce two types of virulence factors: adhesins that promote binding to specific enterocyte receptors for intestinal colonization and enterotoxins responsible for fluid secretion. The best-characterized adhesins are expressed in the context of fimbriae, such as the F4 (also designated K88), F5 (K99), F6 (987P), F17, and F18 fimbriae. Once established in the animal small intestine, ETEC produce enterotoxin(s) that lead to diarrhea. The enterotoxins belong to two major classes: heat-labile toxins that consist of one active and five binding subunits (LT), and heat-stable toxins that are small polypeptides (STa, STb, and EAST1). This review describes the disease and pathogenesis of animal ETEC, the corresponding virulence genes and protein products of these bacteria, their regulation and targets in animal hosts, as well as mechanisms of action. Furthermore, vaccines, inhibitors, probiotics, and the identification of potential new targets by genomics are presented in the context of animal ETEC.
Collapse
Affiliation(s)
- J Daniel Dubreuil
- Faculté de Médecine Vétérinaire, Université de Montréal, Québec J2S 7C6, Canada
| | - Richard E Isaacson
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN 55108
| | - Dieter M Schifferli
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
37
|
Dong WH, Dai CH, Sun L, Wang J, Sun SY, Zhu GQ, Wu SL, Bao WB. Expression of key glycosphingolipid biosynthesis-globo series pathway genes inEscherichia coliF18-resistant andEscherichia coliF18-sensitive piglets. Anim Genet 2016; 47:428-35. [DOI: 10.1111/age.12428] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2016] [Indexed: 12/01/2022]
Affiliation(s)
- W. H. Dong
- College of Animal Science and Technology; Yangzhou University; Yangzhou 225009 China
| | - C. H. Dai
- College of Animal Science and Technology; Yangzhou University; Yangzhou 225009 China
| | - L. Sun
- College of Animal Science and Technology; Yangzhou University; Yangzhou 225009 China
| | - J. Wang
- College of Animal Science and Technology; Yangzhou University; Yangzhou 225009 China
| | - S. Y. Sun
- College of Animal Science and Technology; Yangzhou University; Yangzhou 225009 China
| | - G. Q. Zhu
- College of Animal Science and Technology; Yangzhou University; Yangzhou 225009 China
| | - S. L. Wu
- College of Animal Science and Technology; Yangzhou University; Yangzhou 225009 China
| | - W. B. Bao
- College of Animal Science and Technology; Yangzhou University; Yangzhou 225009 China
| |
Collapse
|
38
|
Hesselager MO, Everest-Dass AV, Thaysen-Andersen M, Bendixen E, Packer NH. FUT1 genetic variants impact protein glycosylation of porcine intestinal mucosa. Glycobiology 2016; 26:607-22. [PMID: 26858341 DOI: 10.1093/glycob/cww009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/21/2016] [Indexed: 12/20/2022] Open
Abstract
A massive use of antibiotics in industrial pig production is a major cause of the rapidly rising bacterial resistance to antibiotics. An enhanced understanding of infectious diseases and of host-microbe interactions has the potential to explore alternative ways to improve pig health and reduce the need for antibiotics. Host-microbe interactions depend on host-expressed glycans and microbe-carrying lectins. In this study, a G > A (nucleotide 307) missense mutation in the porcine α1,2fucosyltransferase 1 gene (FUT1), which has been reported to prevent infections by the common porcine enteric pathogen F18 fimbriated Escherichia coli, provided a unique opportunity to study glycan structures potentially involved in intestinal infections. N- and O-Linked glycans of the intestinal mucosa proteins were characterized in detail using LC-MS/MS. Relative abundances of all glycans were determined and compared between four heterozygous pigs (FUT1-307(A/G)) and four age-matched homozygous pigs from the same 2 litters carrying the missense FUT1 gene constellation (FUT1-307(A/A)). None of the characterized 48 N-linked glycans was found to be regulated by the FUT1 missense mutation, while 11 of the O-linked glycans showed significantly altered abundances between the two genotypes. The overall abundance of H-antigen carrying structures was decreased fivefold, while H-antigen precursors and sialylated structures were relatively more abundant in pigs with the FUT1 missense mutation. These results provide insight into the role of FUT1 on intestinal glycosylation, improve our understanding of how variation in FUT1 can modulate host-microbe interactions, and suggest that the FUT1 genetic variant may help to improve pig gut health.
Collapse
Affiliation(s)
- Marianne O Hesselager
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus 8000, Denmark
| | - Arun V Everest-Dass
- Biomolecular Frontiers Research Centre, Department of Chemistry and Biomolecular Sciences ARC Centre of Excellence in NanoScale BioPhotonics, Macquarie University, Sydney, NSW 2109, Australia
| | | | - Emøke Bendixen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus 8000, Denmark
| | - Nicolle H Packer
- Biomolecular Frontiers Research Centre, Department of Chemistry and Biomolecular Sciences ARC Centre of Excellence in NanoScale BioPhotonics, Macquarie University, Sydney, NSW 2109, Australia
| |
Collapse
|
39
|
Nguyen UV, Coddens A, Melkebeek V, Devriendt B, Goetstouwers T, Poucke MV, Peelman L, Cox E. High susceptibility prevalence for F4 + and F18 +Escherichia coli in Flemish pigs. Vet Microbiol 2016; 202:52-57. [PMID: 26822901 DOI: 10.1016/j.vetmic.2016.01.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/14/2015] [Accepted: 01/18/2016] [Indexed: 10/22/2022]
Abstract
F4 and/or F18 enterotoxigenic Escherichia coli (F4+/F18+ ETEC) are responsible for diarrhea while F18+ verotoxigenic E. coli (F18+ VTEC) cause edema disease in pigs. Both infections can result in severe economic losses, which are mainly the result of the medication, growth retardation and mortality. The susceptibility of piglets to these pathogens is determined by the presence of F4 and F18 receptors (F4R and F18R). Understanding the composition of the susceptibility phenotypes of pigs is useful for animal health and breeding management. This study aimed to determine the prevalence of the F4 ETEC susceptibility phenotypes and F18+E. coli susceptibility among Flemish pig breeds by using the in vitro villous adhesion assay. In this study, seven F4 ETEC susceptibility phenotypes were found, namely A (F4abR+,acR+,adR+; 59.16%), B (F4abR+,acR+,adR-; 6.28%), C (F4abR+,acR-,adR+; 2.62%), D (F4abR-,acR-,adR+; 6.28%), E (F4abR-,acR-,adR-; 24.08%), F (F4abR+,acR-,adR-; 1.05%) and G (F4abR-,acR+,adR-; 0.52%). F4ab and F4ac E. coli showed a stronger degree of adhesion to the intestinal villi (53.40% and 52.88% strong adhesion, respectively), compared to F4ad E. coli (43.46% strong adhesion). Furthermore, the correlation between F4ac and F4ab adhesion was higher (r=0.78) than between F4ac and F4ad adhesion (r=0.41) and between F4ab and F4ad adhesion (r=0.57). For F18+E. coli susceptibility, seven out of 82 pigs were F18R negative (8.54%), but only two of these seven pigs (2.44%) were also negative for F4R. As such, the chance to identify a pig that is positive for a F4 ETEC variant or F18+E. coli is 97.56%. Therefore, significant economic losses will arise due to F4+ and/or F18+E. coli infections in the Flemish pig population due to the high susceptibility prevalence.
Collapse
Affiliation(s)
- Ut V Nguyen
- Laboratory of Immunology, Faculty of Veterinary medicine, Ghent University, Salisburylaan 133, Merelbeke 9820, Belgium.
| | - Annelies Coddens
- Laboratory of Immunology, Faculty of Veterinary medicine, Ghent University, Salisburylaan 133, Merelbeke 9820, Belgium.
| | - Vesna Melkebeek
- Laboratory of Immunology, Faculty of Veterinary medicine, Ghent University, Salisburylaan 133, Merelbeke 9820, Belgium.
| | - Bert Devriendt
- Laboratory of Immunology, Faculty of Veterinary medicine, Ghent University, Salisburylaan 133, Merelbeke 9820, Belgium.
| | - Tiphanie Goetstouwers
- Laboratory of Animal Genetics, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium.
| | - Mario Van Poucke
- Laboratory of Animal Genetics, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium.
| | - Luc Peelman
- Laboratory of Animal Genetics, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, 9820 Merelbeke, Belgium.
| | - Eric Cox
- Laboratory of Immunology, Faculty of Veterinary medicine, Ghent University, Salisburylaan 133, Merelbeke 9820, Belgium.
| |
Collapse
|
40
|
Theuns S, Vyt P, Desmarets LMB, Roukaerts IDM, Heylen E, Zeller M, Matthijnssens J, Nauwynck HJ. Presence and characterization of pig group A and C rotaviruses in feces of Belgian diarrheic suckling piglets. Virus Res 2015; 213:172-183. [PMID: 26677793 DOI: 10.1016/j.virusres.2015.12.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 12/04/2015] [Accepted: 12/04/2015] [Indexed: 10/22/2022]
Abstract
The importance of group A and C rotaviruses (RVA and RVC) in the pathogenesis of diarrhea in Belgian suckling pigs is poorly investigated, and it is not known which strains are circulating in the Belgian suckling pig population. Obtaining better insights in the occurrence of both viral species in the swine population is essential in order to develop accurate diagnostic, therapeutic and prophylactic strategies to protect suckling pigs against diarrhea in a durable manner. In the present study, viral loads of RVA and RVC were quantified in diarrhea samples of suckling piglets less than 2 weeks old, collected on 36 different Belgian farms. On 22 of 36 farms tested (61%), high viral loads of RVA (6.96-11.95 log10 copies/g feces) and/or RVC (5.40-11.63 log10 copies/g feces) were detected. Seventeen RVA isolates were genotyped for their outer capsid proteins VP7 and VP4. Four different G-genotypes (G3, G4, G5 and G9) for VP7 were found together with 4 different P-genotypes (P[6], P[7], P[13] and P[23]) for VP4, in 8 different G/P combinations. All characterized RVC strains belonged to genotype G6 (VP7), except for one strain possessing the G1 genotype. VP4 genes of Belgian RVC strains were genetically heterogeneous, but were classified in the genotype P5. Most rotavirus positive samples also contained Escherichia coli, whereas Clostridium perfringens infections were mainly detected in rotavirus negative samples. Results of the present study offer better insights in the occurrence of RVA and RVC infections in Belgian diarrheic suckling piglets. As a conclusion, routine diagnostic testing for both viral species in cases of diarrhea in suckling pigs is highly recommended. Furthermore, the present findings also offer valuable information for the development of new prophylactic measures against rotavirus. Finally, the relatedness between RVC strains from pigs and other host species is described, and their possible implications in interspecies transmission events are discussed.
Collapse
Affiliation(s)
- Sebastiaan Theuns
- Ghent University, Faculty of Veterinary Medicine, Department of Virology, Parasitology and Immunology, Laboratory of Virology, B-9820, Merelbeke, Belgium.
| | | | - Lowiese M B Desmarets
- Ghent University, Faculty of Veterinary Medicine, Department of Virology, Parasitology and Immunology, Laboratory of Virology, B-9820, Merelbeke, Belgium
| | - Inge D M Roukaerts
- Ghent University, Faculty of Veterinary Medicine, Department of Virology, Parasitology and Immunology, Laboratory of Virology, B-9820, Merelbeke, Belgium
| | - Elisabeth Heylen
- KU Leuven-University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Viral Metagenomics, B-3000, Leuven, Belgium
| | - Mark Zeller
- KU Leuven-University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Viral Metagenomics, B-3000, Leuven, Belgium
| | - Jelle Matthijnssens
- KU Leuven-University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Laboratory of Viral Metagenomics, B-3000, Leuven, Belgium
| | - Hans J Nauwynck
- Ghent University, Faculty of Veterinary Medicine, Department of Virology, Parasitology and Immunology, Laboratory of Virology, B-9820, Merelbeke, Belgium.
| |
Collapse
|
41
|
Baert K, de Geest BG, de Rycke R, da Fonseca Antunes AB, de Greve H, Cox E, Devriendt B. β-glucan microparticles targeted to epithelial APN as oral antigen delivery system. J Control Release 2015; 220:149-159. [DOI: 10.1016/j.jconrel.2015.10.025] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/28/2015] [Accepted: 10/14/2015] [Indexed: 11/16/2022]
|
42
|
Pickard JM, Chervonsky AV. Intestinal fucose as a mediator of host-microbe symbiosis. THE JOURNAL OF IMMUNOLOGY 2015; 194:5588-93. [PMID: 26048966 DOI: 10.4049/jimmunol.1500395] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Fucose is an L-configuration sugar found abundantly in the mammalian gut. It has long been known to be induced there by the presence of bacteria, but only recently have some of the molecular mechanisms behind this process been uncovered. New work suggests that fucose can have a protective role in both gut-centered and systemic infection and inflammation. This review highlights recent studies showing that, in addition to acting as a food source for beneficial gut symbionts, host fucose can suppress the virulence of pathogens and pathobionts. The relevance of gut fucosylation to human diseases also is discussed.
Collapse
Affiliation(s)
- Joseph M Pickard
- Department of Pathology and Committee on Immunology, The University of Chicago, Chicago, IL 60637
| | - Alexander V Chervonsky
- Department of Pathology and Committee on Immunology, The University of Chicago, Chicago, IL 60637
| |
Collapse
|
43
|
Characterization of moose intestinal glycosphingolipids. Glycoconj J 2015; 32:393-412. [PMID: 26104834 PMCID: PMC4515253 DOI: 10.1007/s10719-015-9604-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 05/28/2015] [Accepted: 06/03/2015] [Indexed: 11/04/2022]
Abstract
As a part of a systematic investigation of the species-specific expression of glycosphingolipids, acid and non-acid glycosphingolipids were isolated from three small intestines and one large intestine of the moose (Alces alces). The glycosphingolipids were characterized by binding of monoclonal antibodies, lectins and bacteria in chromatogram binding assays, and by mass spectrometry. The non-acid fractions were complex mixtures, and all had glycosphingolipids belonging to the lacto- and neolactoseries (lactotriaosylceramide, lactotetraosylceramide, neolactotetraosylceramide, Galα3-Lex hexaosylceramide, and lacto-neolactohexaosylceramide), globo-series (globotriaosylceramide and globotetraosylceramide), and isogloboseries (isoglobotriaosylceramide). Penta- and heptaglycosylceramides with terminal Galili determinants were also characterized. Furthermore, glycosphingolipids with terminal blood group O determinants (H triaosylceramide, H type 2 pentaosylceramide, H type 1 penta- and heptaosylceramide) were characterized in two of the moose small intestines, and in the one large intestine, while the third small intestine had glycosphingolipids with terminal blood group A determinants (A tetraosylceramide, A type 1 hexa- and octaosylceramide, A dodecaosylceramide). The acid glycosphingolipid fractions of moose small and large intestine contained sulfatide, and the gangliosides GM3, GD3, GD1a, GD1b, and also NeuGc and NeuAc variants of the Sda ganglioside and the sialyl-globopenta/SSEA-4 ganglioside. In humans, the NeuAc-globopenta/SSEA-4 ganglioside is a marker of embryonic and adult stem cells, and is also expressed in several human cancers. This is the first time sialyl-globopentaosylceramide/SSEA-4 has been characterized in a fully differentiated normal tissue, and also the first time NeuGc-globopentaosylceramide has been characterized.
Collapse
|
44
|
Wu Z, Xia R, Yin X, Huo Y, Zhu G, Wu S, Bao W. Proteomic Analysis of Duodenal Tissue from Escherichia coli F18-Resistant and -Susceptible Weaned Piglets. PLoS One 2015; 10:e0127164. [PMID: 26053838 PMCID: PMC4459693 DOI: 10.1371/journal.pone.0127164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 04/13/2015] [Indexed: 12/31/2022] Open
Abstract
Diarrhea and edema disease in weaned piglets due to infection by Escherichia coli F18 is a leading cause of economic loss in the pig industry. Resistance to E. coli F18 depends on expression of receptors on intestinal epithelial cells, and individual immunity. This study was conducted in Sutai pig E. coli F18-resistant and -susceptible full sib-pair individuals, identified on the basis of resource populations and verification of adhesion assays. The molecular mechanism underlying E. coli F18 resistance was investigated through analysis of the expression of E. coli F18 receptor associated and innate immunity proteins, using proteomics and bioinformatics techniques. Two-dimensional electrophoresis analysis revealed a total of 20 differentially expressed proteins in E. coli F18-resistant and -susceptible groups (10 upregulated and 10 downregulated). A total of 16 differentially expressed proteins were identified by MALDI TOF/TOF mass spectral analysis. According to gene ontology and pathway analysis, differentially expressed proteins were mainly involved in cell adhesion, immune response and other biologically relevant functions. Network analysis of interactions between differentially expressed proteins indicated a likelihood of their involvement in E. coli F18 infection. The expression levels of several important proteins including actin beta (ACTB), vinculin (VCL), heat stress proteins (HSPs) and transferrin (TF) in E. coli F18-resistant and -susceptible individuals were verified by Western blotting, supporting the identification of ACTB, VCL, HSPs and TF as promising candidate proteins for association with E. coli F18 susceptibility.
Collapse
Affiliation(s)
- Zhengchang Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Riwei Xia
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Xuemei Yin
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Yongjiu Huo
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, P. R. China
| | - Shenglong Wu
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Wenbin Bao
- Key Laboratory for Animal Genetics, Breeding, Reproduction and Molecular Design of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- * E-mail:
| |
Collapse
|
45
|
The levels of Brachyspira hyodysenteriae binding to porcine colonic mucins differ between individuals, and binding is increased to mucins from infected pigs with de novo MUC5AC synthesis. Infect Immun 2015; 83:1610-9. [PMID: 25644008 DOI: 10.1128/iai.03073-14] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Brachyspira hyodysenteriae colonizes the pig colon, resulting in mucohemorrhagic diarrhea and growth retardation. Fecal mucus is a characteristic feature of swine dysentery; therefore, we investigated how the mucin environment changes in the colon during infection with B. hyodysenteriae and how these changes affect this bacterium's interaction with mucins. We isolated and characterized mucins, the main component of mucus, from the colon of experimentally inoculated and control pigs and investigated B. hyodysenteriae binding to these mucins. Fluorescence microscopy revealed a massive mucus induction and disorganized mucus structure in the colon of pigs with swine dysentery. Quantitative PCR (qPCR) and antibody detection demonstrated that the mucus composition of pigs with swine dysentery was characterized by de novo expression of MUC5AC and increased expression of MUC2 in the colon. Mucins from the colon of inoculated and control pigs were isolated by two steps of isopycnic density gradient centrifugation. The mucin densities of control and inoculated pigs were similar, whereas the mucin quantity was 5-fold higher during infection. The level of B. hyodysenteriae binding to mucins differed between pigs, and there was increased binding to soluble mucins isolated from pigs with swine dysentery. The ability of B. hyodysenteriae to bind, measured in relation to the total mucin contents of mucus in sick versus healthy pigs, increased 7-fold during infection. Together, the results indicate that B. hyodysenteriae binds to carbohydrate structures on the mucins as these differ between individuals. Furthermore, B. hyodysenteriae infection induces changes to the mucus niche which substantially increase the amount of B. hyodysenteriae binding sites in the mucus.
Collapse
|
46
|
Moonens K, De Kerpel M, Coddens A, Cox E, Pardon E, Remaut H, De Greve H. Nanobody mediated inhibition of attachment of F18 Fimbriae expressing Escherichia coli. PLoS One 2014; 9:e114691. [PMID: 25502211 PMCID: PMC4263667 DOI: 10.1371/journal.pone.0114691] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Accepted: 11/12/2014] [Indexed: 11/19/2022] Open
Abstract
Post-weaning diarrhea and edema disease caused by F18 fimbriated E. coli are important diseases in newly weaned piglets and lead to severe production losses in farming industry. Protective treatments against these infections have thus far limited efficacy. In this study we generated nanobodies directed against the lectin domain of the F18 fimbrial adhesin FedF and showed in an in vitro adherence assay that four unique nanobodies inhibit the attachment of F18 fimbriated E. coli bacteria to piglet enterocytes. Crystallization of the FedF lectin domain with the most potent inhibitory nanobodies revealed their mechanism of action. These either competed with the binding of the blood group antigen receptor on the FedF surface or induced a conformational change in which the CDR3 region of the nanobody displaces the D″-E loop adjacent to the binding site. This D″-E loop was previously shown to be required for the interaction between F18 fimbriated bacteria and blood group antigen receptors in a membrane context. This work demonstrates the feasibility of inhibiting the attachment of fimbriated pathogens by employing nanobodies directed against the adhesin domain.
Collapse
Affiliation(s)
- Kristof Moonens
- Structural & Molecular Microbiology, Structural Biology Research Center, VIB, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Maia De Kerpel
- Structural & Molecular Microbiology, Structural Biology Research Center, VIB, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Annelies Coddens
- Department of Veterinary Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, Merelbeke 9820, Belgium
| | - Eric Cox
- Department of Veterinary Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, Merelbeke 9820, Belgium
| | - Els Pardon
- Structural & Molecular Microbiology, Structural Biology Research Center, VIB, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Han Remaut
- Structural & Molecular Microbiology, Structural Biology Research Center, VIB, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Henri De Greve
- Structural & Molecular Microbiology, Structural Biology Research Center, VIB, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
- * E-mail:
| |
Collapse
|
47
|
Okello E, Moonens K, Erume J, De Greve H. Enterotoxigenic Escherichia coli strains are highly prevalent in Ugandan piggeries but disease outbreaks are masked by antibiotic prophylaxis. Trop Anim Health Prod 2014; 47:117-22. [DOI: 10.1007/s11250-014-0694-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 09/30/2014] [Indexed: 11/24/2022]
|
48
|
Porcine intestinal glycosphingolipids recognized by F6-fimbriated enterotoxigenic Escherichia coli. Microb Pathog 2014; 76:51-60. [PMID: 25241919 DOI: 10.1016/j.micpath.2014.09.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 09/16/2014] [Accepted: 09/17/2014] [Indexed: 11/23/2022]
Abstract
One important virulence factor of enterotoxigenic Escherichia coli is their ability to adhere via fimbrial adhesins to specific receptors located on the intestinal mucosa. Here, the potential glycosphingolipid receptors of enterotoxigenic F6-fimbriated E. coli were examined by binding of purified F6 fimbriae, and F6-expressing bacteria, to glycosphingolipids on thin-layer chromatograms. When intestinal mucosal non-acid glycosphingolipids from single pigs were assayed for F6 binding capacity, a selective interaction with two glycosphingolipids was observed. The binding-active glycosphingolipids were isolated and characterized as lactotriaosylceramide (GlcNAcβ3Galβ4Glcβ1Cer) and lactotetraosylceramide (Galβ3GlcNAcβ3Galβ4Glcβ1Cer). Further binding assays using a panel of reference glycosphingolipids showed a specific interaction between the F6 fimbriae and a number of neolacto core chain (Galβ4GlcNAc) glycosphingolipids. In addition, an occasional binding of the F6 fimbriae to sulfatide, galactosylceramide, lactosylceramide with phytosphingosine and/or hydroxy fatty acids, isoglobotriaosylceramide, gangliotriaosylceramide, and gangliotetraosylceramide was obtained. From the results we conclude that lactotriaosylceramide and lactotetraosylceramide are major porcine intestinal receptors for F6-fimbriated E. coli.
Collapse
|
49
|
Barone A, Benktander J, Teneberg S, Breimer ME. Characterization of acid and non-acid glycosphingolipids of porcine heart valve cusps as potential immune targets in biological heart valve grafts. Xenotransplantation 2014; 21:510-22. [PMID: 25041314 DOI: 10.1111/xen.12123] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 05/28/2014] [Indexed: 02/03/2023]
Abstract
BACKGROUND Although xenotransplantation of vascularized organs/cells has not yet reached the clinic, glutaraldehyde-treated bioprosthetic heart valves (BHV), derived from porcine or bovine tissues, are today used for clinical replacement of diseased heart valves. However, the durability of these valve cusps is limited partly due to the onset of immune responses to the grafts. The xenoantigen-determinant Galα3Gal- and corresponding anti-Gal antibodies have been postulated to in part contribute to BHV damage. However, the presence of other non-Gal carbohydrate antigen determinants as well as the immune response to these non-Gal antigens and the inflammatory response generated by their interaction with the immune system has not been studied. In this study, we have isolated and structurally characterized both non-acid and acid glycosphingolipids from naïve porcine aortic and pulmonary valve cusps. METHODS Total non-acid and acid glycosphingolipids were isolated from porcine aortic and pulmonalis valve cusps of 20 animals. Glycosphingolipid components were structurally characterized by thin-layer chromatography, liquid chromatography-mass spectrometry and binding of monoclonal antibodies and lectins. RESULTS The non-acid glycosphingolipids were characterized as globotetraosylceramide, H-type 2 pentaosylceramide, fucosyl-gangliotetraosylceramide, and Galα3neolactotetraosylceramide. The acid glycosphingolipid fractions had both sulfatide and gangliosides (GM3, GM2, GM1, fucosyl-GM1, GD3 and GD1a), and all gangliosides contained N-acetyl-neuraminic acid. Significantly, the N-glycolyl-neuraminic acid (NeuGc) variant, a major component in many pig organs and to which humans can develop antibodies, was not detected among the gangliosides. CONCLUSIONS Pig valve cusps contain several complex lipid-bound carbohydrate structures that may be targets for the human immune system. Notable, the NeuGc determinant was absent in the cusp gangliosides. This work forms a platform for further characterizing the antibody reactivity of patients with porcine-derived BHV.
Collapse
Affiliation(s)
- Angela Barone
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | | | | | | |
Collapse
|
50
|
Rossi L, Dell'Orto V, Vagni S, Sala V, Reggi S, Baldi A. Protective effect of oral administration of transgenic tobacco seeds against verocytotoxic Escherichia coli strain in piglets. Vet Res Commun 2014; 38:39-49. [PMID: 24249478 DOI: 10.1007/s11259-013-9583-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2013] [Indexed: 11/27/2022]
Abstract
The use of transgenic plants as delivery system for antigenic proteins is attractive for its simplicity and increases likelihood for local immune response at sites of infection. The aim of this study was to evaluate the protective effect of oral administration of tobacco seeds, expressing the FedA, the major protein of the F18 adhesive fimbriae, and B subunit of verocytotoxin, against verocytotoxin-producing E. coli (VTEC) strain in piglets. Forty-three early weaned piglets, were randomly divided into 4 experimental groups: 3 test groups and a control. Treatment groups orally received a bolus, with different dose of tobacco seeds on 0, 1, 2, 14 days post primary administration. After challenge, with 1*10(10) CFU of O138 Escherichia coli strain, piglets showed clinical scores significantly higher in the control group compared to orally immunized groups (P < 0.05) and the latter showed a faster recovery than in CG. In conclusion, oral administration of recombinant tobacco seeds expressing antigenic proteins against VTEC strains can induce a protective effect against challenger strain in piglets.
Collapse
Affiliation(s)
- Luciana Rossi
- Department of Health, Animal Science and Food Safety, Università di Milano, Via Celoria 10, 20133, Milan, Italy,
| | | | | | | | | | | |
Collapse
|